Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis

Synovial fibroblasts (SFs) are key pathogenic drivers in rheumatoid arthritis (RA). Their in vivo activation by TNF is sufficient to orchestrate full arthritic pathogenesis in animal models, and TNF blockade proved efficacious for a high percentage of patients with RA albeit coinducing rare but seri...

Full description

Bibliographic Details
Main Authors: Dimitra Papadopoulou, Fani Roumelioti, Christos Tzaferis, Panagiotis Chouvardas, Anna-Kathrine Pedersen, Filippos Charalampous, Eleni Christodoulou-Vafeiadou, Lydia Ntari, Niki Karagianni, Maria C. Denis, Jesper V. Olsen, Alexios N. Matralis, George Kollias
Format: Article
Language:English
Published: American Society for Clinical investigation 2023-05-01
Series:JCI Insight
Subjects:
Online Access:https://doi.org/10.1172/jci.insight.165024
_version_ 1797634277210324992
author Dimitra Papadopoulou
Fani Roumelioti
Christos Tzaferis
Panagiotis Chouvardas
Anna-Kathrine Pedersen
Filippos Charalampous
Eleni Christodoulou-Vafeiadou
Lydia Ntari
Niki Karagianni
Maria C. Denis
Jesper V. Olsen
Alexios N. Matralis
George Kollias
author_facet Dimitra Papadopoulou
Fani Roumelioti
Christos Tzaferis
Panagiotis Chouvardas
Anna-Kathrine Pedersen
Filippos Charalampous
Eleni Christodoulou-Vafeiadou
Lydia Ntari
Niki Karagianni
Maria C. Denis
Jesper V. Olsen
Alexios N. Matralis
George Kollias
author_sort Dimitra Papadopoulou
collection DOAJ
description Synovial fibroblasts (SFs) are key pathogenic drivers in rheumatoid arthritis (RA). Their in vivo activation by TNF is sufficient to orchestrate full arthritic pathogenesis in animal models, and TNF blockade proved efficacious for a high percentage of patients with RA albeit coinducing rare but serious side effects. Aiming to find new potent therapeutics, we applied the L1000CDS2 search engine, to repurpose drugs that could reverse the pathogenic expression signature of arthritogenic human TNF–transgenic (hTNFtg) SFs. We identified a neuroleptic drug, namely amisulpride, which reduced SFs’ inflammatory potential while decreasing the clinical score of hTNFtg polyarthritis. Notably, we found that amisulpride function was neither through its known targets dopamine receptors D2 and D3 and serotonin receptor 7 nor through TNF–TNF receptor I binding inhibition. Through a click chemistry approach, potentially novel targets of amisulpride were identified, which were further validated to repress hTNFtg SFs’ inflammatory potential ex vivo (Ascc3 and Sec62), while phosphoproteomics analysis revealed that treatment altered important fibroblast activation pathways, such as adhesion. Thus, amisulpride could prove beneficial to patients experiencing RA and the often-accompanying comorbid dysthymia, reducing SF pathogenicity along with its antidepressive activity, serving further as a “lead” compound for the development of novel therapeutics against fibroblast activation.
first_indexed 2024-03-11T12:05:22Z
format Article
id doaj.art-1d4158356be44267b7a2df77d6bc97a8
institution Directory Open Access Journal
issn 2379-3708
language English
last_indexed 2024-03-11T12:05:22Z
publishDate 2023-05-01
publisher American Society for Clinical investigation
record_format Article
series JCI Insight
spelling doaj.art-1d4158356be44267b7a2df77d6bc97a82023-11-07T16:25:35ZengAmerican Society for Clinical investigationJCI Insight2379-37082023-05-0189Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritisDimitra PapadopoulouFani RoumeliotiChristos TzaferisPanagiotis ChouvardasAnna-Kathrine PedersenFilippos CharalampousEleni Christodoulou-VafeiadouLydia NtariNiki KaragianniMaria C. DenisJesper V. OlsenAlexios N. MatralisGeorge KolliasSynovial fibroblasts (SFs) are key pathogenic drivers in rheumatoid arthritis (RA). Their in vivo activation by TNF is sufficient to orchestrate full arthritic pathogenesis in animal models, and TNF blockade proved efficacious for a high percentage of patients with RA albeit coinducing rare but serious side effects. Aiming to find new potent therapeutics, we applied the L1000CDS2 search engine, to repurpose drugs that could reverse the pathogenic expression signature of arthritogenic human TNF–transgenic (hTNFtg) SFs. We identified a neuroleptic drug, namely amisulpride, which reduced SFs’ inflammatory potential while decreasing the clinical score of hTNFtg polyarthritis. Notably, we found that amisulpride function was neither through its known targets dopamine receptors D2 and D3 and serotonin receptor 7 nor through TNF–TNF receptor I binding inhibition. Through a click chemistry approach, potentially novel targets of amisulpride were identified, which were further validated to repress hTNFtg SFs’ inflammatory potential ex vivo (Ascc3 and Sec62), while phosphoproteomics analysis revealed that treatment altered important fibroblast activation pathways, such as adhesion. Thus, amisulpride could prove beneficial to patients experiencing RA and the often-accompanying comorbid dysthymia, reducing SF pathogenicity along with its antidepressive activity, serving further as a “lead” compound for the development of novel therapeutics against fibroblast activation.https://doi.org/10.1172/jci.insight.165024Therapeutics
spellingShingle Dimitra Papadopoulou
Fani Roumelioti
Christos Tzaferis
Panagiotis Chouvardas
Anna-Kathrine Pedersen
Filippos Charalampous
Eleni Christodoulou-Vafeiadou
Lydia Ntari
Niki Karagianni
Maria C. Denis
Jesper V. Olsen
Alexios N. Matralis
George Kollias
Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis
JCI Insight
Therapeutics
title Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis
title_full Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis
title_fullStr Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis
title_full_unstemmed Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis
title_short Repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis
title_sort repurposing the antipsychotic drug amisulpride for targeting synovial fibroblast activation in arthritis
topic Therapeutics
url https://doi.org/10.1172/jci.insight.165024
work_keys_str_mv AT dimitrapapadopoulou repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT faniroumelioti repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT christostzaferis repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT panagiotischouvardas repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT annakathrinepedersen repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT filipposcharalampous repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT elenichristodoulouvafeiadou repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT lydiantari repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT nikikaragianni repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT mariacdenis repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT jespervolsen repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT alexiosnmatralis repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis
AT georgekollias repurposingtheantipsychoticdrugamisulpridefortargetingsynovialfibroblastactivationinarthritis